Abstract
The effect and clinical benefit of P2Y12 receptor antagonists may not be limited to platelet inhibition and the prevention of arterial thrombus formation. Potential additional effects include reduction of the pro‐inflammatory role of activated platelets and effects related to P2Y12 receptor inhibition on other cells apart from platelets. P2Y12 receptor antagonists, thienopyridines and ticagrelor, differ in their mode of action being prodrugs instead of direct acting and irreversibly instead of reversibly binding to P2Y12. These key differences may provide different potential when it comes to additional effects. In addition to P2Y12 receptor blockade, ticagrelor is unique in having the only well‐documented additional target of inhibition, the equilibrative nucleoside transporter 1. The current review will address the effects of P2Y12 receptor antagonists beyond platelets and the protection against arterial thrombosis. The discussion will include the potential for thienopyridines and ticagrelor to mediate anti‐inflammatory effects, to conserve vascular function, to affect atherosclerosis, to provide cardioprotection and to induce dyspnea.
Abbreviations
- ACS
acute coronary syndrome
- AngII
angiotensin II
- ApoE
apoprotein E
- CABG
coronary artery bypass grafting
- CAD
coronary artery disease
- CD39
ectonucleoside triphosphate diphosphohydrolase 1
- CD62P
P‐selectin
- CD73
ecto‐5′‐nucleotidase
- CHD
coronary heart disease
- EC
endothelial cell
- ENT1
equilibrative nucleoside transporter 1
- FBF
forearm blood flow
- FMD
flow‐mediated dilatation
- hsCRP
high sensitive C reactive protein
- NSTEMI
non ST‐elevation myocardial infarction
- PCI
percutaneous coronary intervention
- ROS
reactive oxygen species
- STEMI
ST‐elevation myocardial infarction
- VSMC
vascular smooth muscle cells
Tables of Links
These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2015/16 (a bAlexander et al., 2015a, 2015b).
Introduction
The P2Y12 receptor is a key player in primary haemostasis and in arterial thrombosis and is an established target for antithrombotic drugs including ticlopidine, clopidogrel, prasugrel and ticagrelor.
Ticlopidine, clopidogrel and prasugrel are thienopyridines, which are all prodrugs that require hepatic enzyme‐dependent conversion into an active metabolite that irreversibly bind to P2Y12 receptors. Ticlopidine was introduced in 1980, 21 years before its target receptor was cloned and characterized (Hollopeter et al., 2001). Ticlopidine has been gradually replaced by clopidogrel, which was launched in 1997. Clopidogrel's pharmacodynamic effect varies from 0% to 100% inhibition of ADP‐induced platelet aggregation (Järemo et al., 2002; Aleil et al., 2005). Prasugrel, which was introduced in 2009, has a less variable platelet inhibition driven by a more rapid, complete and consistent generation of its active metabolite, compared with clopidogrel (Wallentin et al., 2008). Ticagrelor, first approved in 2010, is the first member of a new chemical class, cyclopentyltriazolopyrimidines. Ticagrelor is directly acting, reversibly binding to the P2Y12 receptor (van Giezen et al., 2009) and has an additional mode of action, not present for the thienopyridines, as ticagrelor also inhibits cellular adenosine uptake via the equilibrative nucleoside transporter 1 (ENT1) (Armstrong et al., 2014; Cattaneo et al.,2014).
Clopidogrel provides superior clinical benefit compared with aspirin (Gent et al., 1996) and compared with placebo when given in addition to aspirin in acute coronary syndrome (ACS) patients (Yusuf et al., 2001). Both prasugrel and ticagrelor provide superior clinical benefit compared with clopidogrel when given on top of aspirin in ACS patients (Wiviott et al., 2007; Wallentin et al., 2009). In addition, ticagrelor have shown superior clinical benefit compared with placebo when given on top of aspirin in patients with a prior myocardial infarction (Bonaca et al., 2015). True for all antiplatelet agents is that they do increase the risk of bleeding and that the clinical benefit in the studies listed previously is accompanied with an increased bleeding risk including life‐threatening bleeds. Thus, careful consideration of the benefit–risk ratio for each patient is needed to optimize the anti‐platelet therapy in terms of drugs to use and length of treatment on an individual level.
In addition to these already launched P2Y12 receptor antagonists, there are two other non‐thienopyridine P2Y12 receptor antagonists that have been tested in patients. Cangrelor, an ATP analogue, is a direct acting and reversibly binding P2Y12 receptor antagonist. However, unlike ticagrelor, cangrelor is intended for acute treatment only, as the compound is short acting (t1/2 3–6 min) and not orally administered (Lhermusier et al., 2015). Acute treatment with cangrelor is superior to standard clopidogrel treatment when given as a bolus and infusion to patients undergoing urgent or elective percutaneous coronary interventions (PCI) (Bhatt et al., 2013). Cangrelor has just recently been approved as an adjunct to PCI. Finally, elinogrel is also a direct acting reversibly binding P2Y12 receptor antagonist, but belongs to a different chemical class from ticagrelor and cangrelor. Elinogrel was intended both for p.o. and i.v. administration, but development was stopped in Phase II clinical trials.
This review will focus on the effects of ticagrelor and the thienopyridines and will summarize the evidence for effects of P2Y12 receptor antagonists beyond platelet inhibition in the cardiovascular system.
Drug exposure
The pharmacodynamic effect of P2Y12 receptor antagonists, on or off target, is related to the exposure of the active drug. The active metabolites of the thienopyridines can only be detected in the systemic circulation for about 4 h following intake of the prodrug and then only at levels below their in vitro IC50 values, determined as the concentration providing 50% inhibition of ADP‐induced platelet aggregation (Figure 1B). One can therefore argue that the thienopyridines have limited or no systemic exposure, and one may hypothesize that the platelet inhibition by thienopyridines mainly takes place in the hepatic circulation where the active metabolites are formed and therefore likely present at higher concentrations. Because of their irreversible mode of action, however, thienopyridines can overcome the shorter duration of action of their active metabolite so that they still can provide platelet inhibition over 24 h.
Figure 1.
Illustration of plasma exposure of active drug over 24 h following ticagrelor 90 mg twice daily (A) and clopidogrel 75 mg once daily or prasugrel 10 mg once daily (B). In A, ticagrelor (dotted red line), ARC124910XX (dashed red line) and the sum of ticagrelor and AR‐C124910XX (solid red line). In B, clopidogrel active metabolite (AM) (purple line) and prasugrel‐AM (green line). Illustration is based on published patient exposure data (Wallentin et al., 2008; Storey et al., 2007). Dotted black lines represent IC50 values, using ADP‐induced light transmission aggregometry in human PRP in vitro. In A, IC50 is the mean of ticagrelor and AR‐C124910XX data presented in Table 1. In B, clopidogrel‐AM and prasugrel‐AM IC50 data is from Sugidachi et al., 2007.
In contrast to thienopyridines, ticagrelor does not require hepatic enzyme‐dependent conversion into an active metabolite. That said ticagrelor has a major circulating metabolite, AR‐C124910XX, with plasma exposure of 30–40% (Storey et al., 2007) and similar potency versus P2Y12 as the parent ticagrelor (Table 1). Ticagrelor is given twice a day, which provides a biphasic increase in the plasma concentration of ticagrelor and AR‐C124910XX over 24 h (Figure 1A). Given that both ticagrelor and its metabolite are active and reversibly binding to the platelet P2Y12 receptor, the main pharmacodynamic effect, platelet inhibition, is directly related to the combined exposure of ticagrelor and AR‐C124910XX in the systemic circulation (Husted et al., 2012). As predicted from the in vitro potency data, a high and continuous platelet inhibition is achieved in patients as the plasma exposure of ticagrelor is well above its in vitro IC50 throughout 24 h (Figure 1). The presence of an active drug in the systemic circulation throughout 24 h provides what can be referred to as a systemic potential.
Table 1.
The mean concentrations that gave half‐maximum inhibition (IC50) of ticagrelor and AR‐C124910XX with regards to receptor binding, receptor signalling and ADP‐induced platelet aggregation in whole blood and platelet‐rich plasma (PRP) is listed together with the binding affinity (Ki) at platelet P2Y12 receptors
In vitro inhibition of human P2Y12 receptor radioligand binding and 2MeSADP‐induced receptor signalling | |||
---|---|---|---|
Receptor binding, IC50 μM, (n) | Ki, nM, (n) | Receptor signalling, IC50 μM, (n) | |
Ticagrelor | 0.01 (63) | 2.0 (8) | 0.07 (98) |
AR‐C124910XX | 0.06 (2) | 2.5 (2) | 0.05 (2) |
In vitro inhibition of ADP‐induced platelet aggregation in human whole blood and PRP | |||
---|---|---|---|
Whole blood, IC50 μM, (n) | PRP, IC50 μM, (n) | ||
Ticagrelor | 0.24 (4) | 0.40 (8) | |
AR‐C124910XX | 0.17 (4) | 0.13 (8) |
For receptor binding the displacement of [125I]AZ11931285, a radio‐ligand that binds to the same site on the receptor was used, and for receptor signalling, the inhibition of 2MeSADP‐induced P2Y12 receptor signalling using a 35S‐GTPγS assay was evaluated. Both assays used membranes from human P2Y12 receptor transfected cells. ADP‐induced platelet aggregation was evaluated in whole blood with impedance aggregometry using Multiplate (Dynabyte, Munich, Germany) and in PRP with light transmission aggregometry using PAP8 (BioData Corporation, PA, USA). The data is unpublished except for the Ki data for ticagrelor (Springthorpe et al., 2007). Overall data indicate similar potency at P2Y12 receptors for ticagrelor and AR‐C124910XX.
This systemic potential means that ticagrelor apart from platelet P2Y12 receptors should be able to reach additional targets (non‐platelet P2Y12 receptors and the equilibrative nucleoside transporter 1, ENT1 for ticagrelor) expressed systemically.
ENT1 inhibition by ticagrelor
Adenosine is formed from ATP and ADP released locally at sites of ischaemia, tissue damage and inflammation. The t1/2 of adenosine in the circulation is a matter of seconds due to its rapid cell uptake and intracellular metabolism. Ticagrelor inhibits the ENT1 transporter and thereby reduces the cellular uptake of adenosine resulting in its prolonged local t1/2 and extracellular presence (Nylander et al., 2013; Armstrong et al., 2014). Ticagrelor has been shown to increase the plasma levels of adenosine in ACS patients (Bonello et al., 2013) and to augment adenosine‐induced physiological responses, including increases in coronary blood flow (Alexopoulos et al., 2013; Wittfeldt et al., 2013), platelet inhibition (Nylander et al., 2013), neutrophil migration (Alsharif et al., 2015) and the sensation of dyspnea (Wittfeldt et al., 2013). The additional mode of action, ENT1 inhibition and its potential clinical relevance has recently been reviewed (Cattaneo et al., 2014). Importantly, and unlike P2Y12 receptors, only ticagrelor and not AR‐C124910XX significantly inhibits ENT1 (Armstrong et al., 2014). In addition to ENT1 inhibition, ticagrelor induced ATP release from human erythrocytes in vitro (Öhman et al., 2012); it remains unknown whether this effect of ticagrelor on erythrocytes occurs also in vivo. If present, both mechanisms would work together to further enhance the local concentration of adenosine as ATP is rapidly converted into AMP via the ectonucleoside triphosphate diphosphohydrolase 1 (CD39), with subsequent conversion into adenosine via the ecto‐5′‐nucleotidase (CD73) (Cattaneo et al., 2014).
Juvenile platelets
Patients with increased platelet turnover have higher levels of immature platelets, and an increase in the immature platelet count has been found to be associated with worse cardiovascular outcome (Cesari et al., 2013; Ibrahim et al., 2014) supporting the hypothesis that these juvenile platelets are more pro‐thrombotic. A continuous systemic exposure of an active compound, as true for ticagrelor, should theoretically inhibit newly formed, juvenile platelets equally well as the old platelets. This should not be the case for the thienopyridines as the platelets formed after elimination of the active metabolites from the circulation will not be inhibited until the next dose of prodrug on the next day. In support, high immature platelets count has been found associated with lower anti‐platelet response to clopidogrel (Ibrahim et al., 2012). A rat study has shown that the recovery of platelet function after ticagrelor administration differs from that after clopidogrel treatment (Kuijpers et al., 2011). Following ticagrelor administration, all platelets (old and newly formed) were inhibited, and their function gradually recovered over time in parallel with drug elimination. Following clopidogrel treatment, there was a gradually emerging subpopulation of uninhibited platelets, probably juvenile platelets. This difference in the platelet inhibition properties is masked by conventional platelet aggregation tests, which evaluate the net response of the total platelet population, but was revealed by thrombus formation measurement under flow where the juvenile un‐inhibited platelets, formed at later time points after clopidogrel treatment, appeared to support thrombus formation (Kuijpers et al., 2011). In another study a mixture of uninhibited and prasugrel‐inhibited platelets was studied to model the role of juvenile platelets formed each day. Twenty percent prasugrel‐inhibited plus 80% uninhibited platelets were used to mimic diabetics that have a high platelet turnover with an approximate platelet lifespan of 5 days (thus about 20% new platelets should be formed each day). By imaging of ADP‐induced aggregates of these mixtures, the authors showed that uninhibited platelets supported a core of aggregated platelets, which was surrounded by prasugrel‐inhibited platelets (Hoefer et al., 2015; Gurbel and Tantry, 2015). Another recent study explored platelet function after discontinuation of prasugrel in ACS patients. They found that prasugrel discontinuation resulted in the formation of an emerging subpopulation of ADP‐responsive juvenile platelets, which contributed to platelet aggregation and thrombus formation under flow (Baaten et al., 2015). Finally, a clinical study explored platelet function in ACS patients on ticagrelor or prasugrel treatment (Bernlochner et al., 2015). They found that the number of juvenile platelets (immature platelet count) correlated with platelet aggregation in patients treated with prasugrel, but not in those receiving ticagrelor, indicating that juvenile platelets contribute to increased platelet aggregation in prasugrel‐treated, but not with ticagrelor‐treated patients. Flow cytometry experiments confirmed that the juvenile platelets were less inhibited in prasugrel, compared with ticagrelor‐treated patients especially late after drug intake (1 h before next dose).
Non‐platelet P2Y 12 receptors
Historically, P2Y12 receptors have been seen as platelet‐specific. More recently, it has become clear that many cells express P2Y12 receptors including endothelial cells (EC) (Simon et al., 2002; Shanker et al., 2006), vascular smooth muscle cells (VSMC) (Wihlborg et al., 2004; Satonaka et al., 2015), dendritic cells (Ben Addi et al., 2010), leukocytes (Wang et al., 2004; Diehl et al., 2010) and neurons (Kawaguchi et al., 2015). The roles of these non‐platelet P2Y12 receptors are poorly explored, but data are emerging (Gachet, 2012).
Activation of P2Y12 receptors expressed on EC decreased intracellular cAMP content (Simon et al., 2002). EC cAMP content, via activation of PKA, was responsible for endothelial barrier function (Lum et al., 1999), which was disturbed under pathophysiological conditions including increased inflammation (Aslam et al., 2014) or hypoxia/reoxygenation (Aslam et al., 2013). Disruption of endothelial barrier function was attenuated by elevation of EC cAMP levels (Aslam et al., 2013, 2014) which might occur following blockade of P2Y12 receptors (Simon et al., 2002).
The VSMC P2Y12 receptors have been shown to induce vasoconstriction when activated by ADP (Wihlborg et al., 2004). Ticagrelor inhibited this ADP‐induced VSMC vasoconstriction in vitro whereas in vivo thienopyridine treatment had no effect, most likely because of no or low presence of the active metabolites at the site of the target (Högberg et al., 2010; Grzesk et al., 2012) (Figure 1) The active metabolite of prasugrel, when added in vitro, inhibited the P2Y12 receptor agonist 2‐methylthio‐ADP‐induced VSMC IL‐6 expression and mitogenesis (Rauch et al., 2010). The clinical relevance of non‐platelet P2Y12 receptors and their inhibition, including VSMC P2Y12, is unknown.
A hypothesis has been proposed that non‐platelet, potentially neuronal P2Y12 receptors, could be the mediators of drug‐induced dyspnea observed in patients (Cattaneo and Faioni, 2012). This hypothesis is supported by the observation that all direct acting and reversibly binding P2Y12 receptor antagonists evaluated in patients (ticagrelor, cangrelor and elinogrel) increased the incidence of dyspnea. In the PLATO study, the incidence of dyspnea was 13.8% versus 7.8% in ticagrelor‐treated and clopidogrel‐treated patients and in the PEGASUS study, 18.9%, 15.8% and 6.4% experienced dyspnea when 90 mg ticagrelor, 60 mg ticagrelor or placebo was given on top of aspirin (Wallentin et al., 2009; Bonaca et al., 2015). In the INNOVATE‐PCI study, elinogrel induced 12.3% dyspnea versus 3.8% for clopidogrel (Welsh et al., 2012). In studies with cangrelor, the incidence of dyspnea is much lower, probably because of the short duration of treatment, but still highly significant (Bhatt et al., 2009; Bhatt et al., 2013; Harrington et al., 2009). The characteristics of ticagrelor‐induced dyspnea in the PLATO study has been published, and the authors concluded that it was usually mild or moderate in intensity and not associated with differences in efficacy or safety outcomes in the study (Storey et al., 2011). As adenosine is known to induce dyspnea (Burki et al., 2005), an alternative hypothesis to explain ticagrelor‐induced dyspnea is increased extracellular adenosine levels via ENT1 inhibition. Indeed, as mentioned, ticagrelor enhanced adenosine‐induced dyspnea in healthy subjects (Wittfeldt et al., 2013). However, unlike ticagrelor, cangrelor and elinogrel do not inhibit cellular adenosine uptake (Armstrong et al., 2014), but still increased the incidence of dyspnea. Thus, the adenosine hypothesis does not explain what may be a class effect of these compounds (Cattaneo et al., 2014). As dyspnea is a sensation, it is hard to explore the mechanism responsible in animal models why dyspnea needs to be explored in patients that can express their level of breathlessness. Currently, a clinical trial is ongoing to explore the hypothesis that ticagrelor‐induced dyspnea is mediated via adenosine. In this study, ticagrelor patients experiencing dyspnea will be randomized to receive caffeine (an adenosine receptor antagonist) or placebo for 1 week, and the primary end point is change in visual analogue scale AUC for dyspnea (Lindholm et al., 2015).
In summary, the pharmacokinetic and pharmacodynamic properties of available P2Y12 receptor antagonists have a number of key differences. This provides different potential to inhibit systemically expressed non‐platelet P2Y12 receptors and off targets, such as inhibition of ENT1 in the case of ticagrelor. Furthermore, 24 h plasma exposure of active compound is required to provide continuous platelet inhibition of old and newly formed platelets alike.
Effects on inflammation
It is well known that platelets play a role in inflammatory responses (see Gros et al., 2015; Nording et al., 2015; Thomas and Storey, 2015). Platelets store pro‐inflammatory cytokines that are released when platelets are activated. Also microRNAs, small non‐coding RNAs, such as micro RNA‐223, RNA‐126, RNA‐21, RNA‐24 and RNA‐197 are known to be present in platelets and are secreted in exosomes and/or microvesicles upon platelet activation (Hulsmans and Holvoet, 2013; Fuentes et al., 2015). These micro RNAs, once taken up by cells, can further trigger inflammatory processes and increase atherosclerosis and angiogenesis (see Fuentes et al., 2015). In addition, activated platelets translocate adhesion receptors, for example, CD62P (P‐selectin) and CD40L from their α granules to the plasma membrane. CD62P will bind to P‐selectin glycoprotein ligand‐1 on leukocytes and create platelet–leukocyte aggregates and trigger leukocyte activation. CD40L will bind to CD40 on a number of cells including EC and trigger their activation. As a result of leukocyte and EC activation, more pro‐inflammatory cytokines will be released, and more adhesion receptors expressed at the site of inflammation that further amplifies the inflammatory process (Figure 2). Thus, inhibition of platelet activation will attenuate the pro‐inflammatory capacity of platelets. All anti‐platelet drugs should have this effect to different extent depending on the importance of the target and the level of target inhibition.
Figure 2.
Illustration of the role of platelets in amplification of inflammation via interactions with leukocytes and endothelial cells and the release of cytokines from all cells involved. Platelets store pro‐inflammatory cytokines that are released when platelets are activated. In addition, activated platelets translocate adhesion receptors, for example, CD62P (P‐selectin) and CD40L from their α granules to the plasma membrane. CD62P will bind to P‐selectin glycoprotein ligand‐1 on leukocytes and create platelet–leukocyte aggregates and trigger leukocyte activation. CD40L will bind to CD40 on a number of cells including endothelial cells (EC) and trigger their activation. As a result of leukocyte and EC activation, more pro‐inflammatory cytokines will be released, and more adhesion receptors expressed at the site of inflammation that further amplifies the inflammatory process. This pro‐inflammatory role of platelets can have important implications in inflammatory processes including atherosclerosis.
Thienopyridines
In line with the reasoning outlined above , inhibition of P2Y12 receptors in vitro by clopidiogrel's active metabolite reduced P‐selectin expression, platelet‐polymorphonuclear leukocyte adhesion and production of reactive oxygen species (ROS) by polymorphonuclear leukocytes (Evangelista et al., 2005). In rabbits, vascular inflammation following myocardial ischaemia/reperfusion was reduced by clopidogrel associated with preserved NO synthase (NOS) expression following clopidogrel treatment, compared with placebo (Molero et al., 2005).
The active metabolite of prasugrel yielded a concentration‐dependent inhibition of platelet aggregation, soluble CD40L (sCD40L) release and platelet–leukocyte co‐aggregate formation in human blood in vitro (Judge et al., 2008). The latter finding was confirmed by others in human blood samples (Frelinger et al., 2007) and extended to the inhibition of agonist‐stimulated platelet–monocyte adhesion in blood from prasugrel treated mice (Totani et al., 2012) (see Schrottmaier et al., 2015, for the platelet–leukocyte/monocyte interaction). Apart from inhibiting platelet–leukocyte and platelet–monocyte adhesions, prasugrel reduced TNF‐α synthesis and increased NO metabolites in endotoxin‐treated mice in vivo (Totani et al., 2012). Interestingly, the anti‐inflammatory actions of the active metabolite of prasugrel was likely derived from direct targeting of neutrophils isolated from human blood and was P2Y12 receptor independent (Liverani et al., 2013). This has been recently reviewed by Thomas and Storey (2015.
Switching aspirin (75 mg·day−1) to clopidogrel (75mg·day−1) reduced the concentrations of some inflammatory markers (in particular high sensitive C reactive protein (hsCRP), IL‐6 and CD40L) in diabetic patients with remaining high platelet reactivity (Rosiak et al., 2013). In contrast, in patients with coronary heart disease (CHD), no between‐group differences in circulating markers of inflammation after 1 year treatment with clopidogrel (75 mg·day−1) compared with aspirin (160 mg·day−1) was evident, but both treatments reduced the levels of TNF‐α (Solheim et al., 2006). Adding clopidogrel on top of aspirin further reduced platelet CD40L expression (Quinn et al., 2004) and reduced sCD40L (Azar et al., 2006). Clopidogrel abolished the influence of leukocytes on platelets (Diehl et al., 2010) and decreased the elevated platelet–monocyte and platelet–neutrophil conjugates in the blood of ACS patients and prevented their agonist‐triggered formation ex vivo (Xiao and Théroux, 2004), while such an effect was not seen in patients with non ST‐elevation myocardial infarction (NSTEMI) at 4 weeks of clopidogrel treatment (Palmerini et al., 2010). Also, the plasma level of hsCRP was reduced in patients on additional treatment with clopidogrel, but not in patients on placebo (Heitzer et al., 2006; Chen et al., 2006). In ACS patients undergoing PCI, hsCRP increased significantly at day 5 in clopidogrel naïve patients, but was reduced in patients on clopidogrel (Vivekananthan et al., 2004). Similarly, in low clopidogrel responders, hsCRP increased 7 days post PCI while in high responders, hsCRP remained within the reference intervals (Malek et al., 2007). In patients receiving long‐term clopidogrel therapy compared with the clopidogrel‐naive group, lower levels of selected inflammation markers (IL, TNF‐α and hsCRP) were achieved (Antonino et al., 2009).
In support of the effects of clopidogrel, in patients undergoing PCI, the plasma levels of sCD40L, hsCRP and IL‐6 after 6 month were higher in patients with clopidogrel resistance than in those with normal clopidogrel responsiveness (Ge et al., 2012; see also Woo et al., 2014). Clopidogrel withdrawal was associated with an increase in platelet and inflammatory biomarkers in diabetic patients, again supporting anti‐inflammatory affects coupled to P2Y12 receptor antagonism (Angiolillo et al., 2006). While the vast majority of studies with clopidogrel have demonstrated an anti‐inflammatory effect, a few studies have failed to show a reduction in hsCRP (Table 2). Clopidogrel therapy also had no consistent effect on EC function, arterial stiffness and oxidative stress markers (Ostad et al., 2011), or the number of circulating progenitor cells (Ramadan et al., 2014); see Husted et al., 2010; Willoughby et al., 2014). Angiogenic cytokines influence vessel injury, and platelets represent a disposable circulating pool of angiogenic molecules. In 28 healthy male volunteers treated for 7 days with clopidogrel, the level of circulating angiogenic factor (VEGF, placenta growth factor and stromal cell‐derived factor‐1) remained unchanged (Smadja et al., 2012).
Table 2.
Studies on clopidogrel describing its direct platelets effects, effects on platelet‐leukocyte interaction, effects on inflammation or vascular reactivity
Clopidogrel | Patients | Aspirin | Platelet (sCD40L, CD62) | Platelet‐leukocyte interaction | Inflammation (hsCRP, TNF, IL) | Oxidative stress | Vascular reactivity (FMD, FBF) | Reference |
---|---|---|---|---|---|---|---|---|
24 h 5 days | ACS PCI (n = 833) | Yes | NA | NA | unchanged reduced | NA | NA | Vivekananthan et al., 2004 |
24 h | ACS (n = 23) | Yes | reduced | reduced | NA | NA | NA | Xiao and Théroux, 2004 |
5 days | PCI (n = 79) | Yes | reduced | NA | unchanged | NA | NA | Quinn et al., 2004 |
24 h | ACS (n = 103) | Yes | unchanged | NA | unchanged | NA | NA | Montalescot et al., 2006 |
24 h | PCI (n = 60) | Yes | NA | NA | TNF reduced CRP unchanged | NA | NA | Gurbel et al., 2006 |
1–4 weeks | ACS (n = 115) | Yes | NA | NA | reduced | NA | NA | Chen et al., 2006 |
5 weeks | CAD (n = 103) | Yes | reduced | NA | reduced | NA | increased | Heitzer et al., 2006 |
8 weeks | CAD (n = 73) | Yes | reduced | NA | unchanged | NA | NA | Azar et al., 2006 |
12 months withdrawal | Diabetes PCI (n = 54) | Yes | increased (upon withdrawal) | NA | Increased (upon withdrawal) | NA | NA | Angiolillo et al., 2006 |
12 month | CHD (n = 101) | No | unchanged | NA | TNF reduced CRP unchanged | NA | NA | Solheim et al., 2006 |
responder versus non‐responder 7 days | STEMI, PCI (n = 34) | Yes | NA | NA | decreased | NA | NA | Malek et al., 2007 |
22 hours | CAD (n = 58) | Yes | NA | NA | NA | NA | increased | Warnholtz et al., 2008 |
Naive versus 6 month | PCI (n = 110) | Yes | NA | NA | reduced | NA | NA | Antonino et al., 2009 |
1–4 weeks | STEMI PCI (n = 54) | Yes | NA | unchanged | reduced | NA | NA | Palmerini et al., 2010 |
4 weeks | NTSE‐ACS (n = 990) | Yes | unchanged | NA | unchanged | unchanged | NA | Husted et al., 2010 |
28 days | CAD (n = 120) | Yes | NA | NA | unchanged | unchanged | unchanged | Ostad et al., 2011 |
6 months responder versus non‐responder | PCI (n = 352) | Yes | reduced | NA | reduced | NA | NA | Ge et al., 2012 |
8 weeks | CAD, diabetes (n = 234) | switching to clopidogrel | reduced | NA | reduced | NA | NA | Rosiak et al., 2013 |
6 weeks | CAD (n = 41) | Yes | reduced | NA | unchanged | unchanged | unchanged | Ramadan et al., 2014 |
3 month | CAD (n = 35) | Yes | NA | NA | NA | unchanged | increased | Willoughby et al., 2014 |
498 days | CAD (n = 91) | Yes | reduced | NA | reduced | NA | increased | Woo et al., 2014 |
4 weeks Responder vs. Non‐responder | CAD, PCI (n = 108) | Yes | NA | NA | NA | Na | increasd | Siasos et al., 2015 |
In patients with sickle cell disease, prasugrel reduced the number of platelets adhering to monocytes and neutrophils (Jakubowski et al., 2014). While the in vivo levels of platelet–monocyte aggregates did not differ between patients receiving clopidogrel or prasugrel treatment, the levels were significantly higher in blood from patients treated with clopidogrel as compared with prasugrel following in vitro platelet activation (Gremmel et al., 2013). However, such difference was not seen after 1 month treatment in another study in patients with stable CHD (Braun et al., 2008). An anti‐inflammatory response of prasugrel was also tested in asthmatic patients, randomly and blindly allocated to prasugrel or placebo for 15 days. In this study, the provocative dose of mannitol causing a 15% drop in FEV1, tended to increase from 142 to 187 mg, after prasugrel, but did not change after placebo treatment. These results suggest that P2Y12 inhibition may reduce the bronchial inflammatory burden (Lussana et al., 2015). More recently, prasugrel, when given to healthy subjects, inhibited the increased release of pro‐inflammatory cytokines when isolated T‐cells from treated subjects was stimulated in the presence of isolated platelets ex vivo (Johnston et al., 2015).
Ticagrelor
In a mouse caecal ligation and puncture model of polymicrobial sepsis, ticagrelor reduced formation of neutrophil–platelet aggregates and lung damage driven by neutrophil recruitment (Rahman et al., 2014). In addition to anti‐inflammatory effects mediated by inhibition of platelet P2Y12 receptors, adenosine is a potent modulator of inflammatory responses. The ability of ticagrelor to provide anti‐inflammatory effects via adenosine has been shown in vitro as ticagrelor enhanced adenosine‐induced neutrophil migration (Alsharif et al., 2015).
Clinically, a post hoc analysis of the PLATO trial revealed that ticagrelor compared with clopidogrel was associated with a lower morbidity and mortality related to pulmonary infection and sepsis (Storey et al., 2014). Thus, an anti‐inflammatory effect may have contributed to the overall mortality benefit seen in the PLATO trial. However, such a beneficial effect was not seen in the recent PEGASUS trial where numerically more deaths related to infection were seen with 90 mg relative to 60 mg ticagrelor and placebo (Bonaca et al., 2015). The reason for the discrepancy between the trials is unknown but may be related to the different patient populations studied. For instance, an anti‐inflammatory effect may have been especially important in patients with coronary artery bypass grafts (CABG), who are likely to be more prone to infection. In the PLATO trial, ticagrelor reduced mortality by 50% compared with clopidogrel in CABG patients (Held et al., 2011; Varenhorst et al., 2012]. In the more stable patients evaluated in the PEGASUS trial, less patients should have undergone CABG, which could in part explain the difference in infection or sepsis related mortality.
As ticagrelor confers higher and more consistent P2Y12 receptor inhibition compared with clopidogrel, ticagrelor should be more effective with regards to any protective effects mediated by P2Y12 receptor inhibition. Indeed, ticagrelor and clopidogrel reduced the release of the pro‐inflammatory cytokine TNF‐α, but only ticagrelor also significantly reduced IL‐6 in a model of human sepsis (Thomas et al., 2014). However, in the PLATO trial, there were no significant differences in inflammatory biomarkers hsCRP, IL‐6, myeloperoxidase or sCD40L in patients treated with ticagrelor versus clopidogrel (Husted et al., 2010). This discrepancy may be related to the time between biomarker sampling, occurrence of the potential inflammatory trigger and timing of last drug dose, which was not captured in the PLATO trial. Currently, the potential effects of ticagrelor on mortality is being assessed in patients with pneumonia (Ticagrelor in Severe Community Acquired Pneumonia (TCAP), ClinicalTrials.gov. Identifier: NCT01998399). The effects of P2Y12 receptor antagonists on inflammation and immunity have been recently reviewed (Thomas and Storey, 2015).
In summary, the vast majority of studies show that inhibition of platelet P2Y12 receptor reduced the amplification of inflammatory responses induced by activated platelets. The level of the anti‐inflammatory effect appears to depend on the level of P2Y12 receptor inhibition although some P2Y12 receptor‐independent effects have been described. Clinically, extensive P2Y12 receptor blockade may result in reduced morbidity and mortality in patients with increased pro‐inflammatory burden.
Effects on vascular function
Thienopyridines
Clopidogrel released NO from cultured EC independently from its anti‐platelet action, and thienopyrimidinone congeners that were devoid of anti‐platelet action also stimulated coronary endothelium to release NO (Jakubowski et al., 2005). Similarly, clopidogrel caused relaxation of arterial tissues and influenced VSMC proliferation directly without hepatic biotransformation in vitro. The effect was independent of endothelium, β‐adrenoceptor and purinergic P2 receptors (Froldi et al., 2011).
In experimental disease models in vivo, endothelium‐dependent relaxation upon ACh stimulation was reduced in rats made hypertensive by angiotensin (Ang) II‐infusion, and clopidogrel treatment was effective in improving endothelial function. Clopidogrel also prevented vascular remodelling, shown by augmented media thickness in aortas from Ang II‐hypertensive rats (Giachini et al., 2014). Similarly, in chronic heart failure rats with reduced NO‐mediated vasorelaxation, clopidogrel treatment improved the vasorelaxation. The effect of clopidogrel was associated with an enhanced phosphorylation of Akt and endothelial NOS (Schäfer et al., 2011).
In patients with symptomatic coronary artery disease (CAD), clopidogrel improved endothelial NO bioavailability and diminished biomarkers of oxidant stress (Heitzer et al., 2006). A higher clopidogrel maintenance dose (150 mg·day−1) was associated with a greater improvement of endothelial function compared with the standard 75 mg·day−1 (Patti et al., 2011). Clopidogrel reduced endothelial injury (assessed as number of circulating EC) during PCI (Bonello et al., 2010) and improved vascular reactivity, either P2Y12 receptor‐dependent (Hamilos et al., 2011; Siasos et al., 2015) or receptor‐independent (Warnholtz et al., 2008; Willoughby et al., 2014). However, such beneficial effect of clopidogrel on vascular reactivity was not seen in all clinical studies, and the beneficial effect of clopidogrel on flow‐mediated brachial artery dilation was not sustained after long‐term clopidogrel treatment (28 days) (Ostad et al., 2011) (Table 2).
Ticagrelor
Ticagrelor increased eNOS activation and COX2 protein expression in TNF‐α stimulated human aortic EC in vitro by a yet unknown mechanism (Reiner et al., 2013). Also, in vivo treatment with ticagrelor increased eNOS activation and both the activation and expression of COX2 in a rat model of ischaemia/reperfusion injury. These effects of ticagrelor were dependent on adenosine receptor activation with no effect of clopidogrel in spite of dosing to similar levels of P2Y12 receptor blockade as ticagrelor (Nanhwan et al., 2014). In a prospective randomized clinical study, ticagrelor increased the number of circulating endothelial progenitor cell compared with clopidogrel in ACS patients (Bonello et al., 2015). Finally, in a non‐randomized study, ACS patients on ticagrelor treatment but not on clopidogrel or prasugrel treatment had improved peripheral endothelial function as evaluated with the EndoPAT system (Torngren et al., 2013). An ongoing study evaluates the acute and chronic effects of clopidogrel, prasugrel and ticagrelor on parameters of vascular and platelet function in ACS patients (Schnorbus et al., 2014).
In summary, thienopyrimidinones devoid of antiplatelet action increase NO production. Clopidogrel has been proven beneficial regarding EC/vascular function, part of it being P2Y12 receptor‐independent. Similarly, ticagrelor increased eNOS but also COX2 expression and activity and, unlike clopidogrel and prasugrel, appears to improve peripheral EC function in ACS patients.
Effects on atherosclerosis
Many risk factors contribute to the accelerated development of atherosclerotic lesions; among them are hypertension, hypercholesterolemia and diabetes. In apoprotein E (ApoE) knockout mice, having a disturbed cholesterol metabolism, platelets adhered to the vascular endothelium of the carotid artery before the development of atherosclerotic lesions (Dong et al., 2000; Massberg et al., 2002). Platelet adhesion to the endothelium coincided with inflammatory gene expression and preceded atherosclerotic plaque invasion by leukocytes (Massberg et al., 2002). Once activated, platelets produce ROS via NADPH oxidases (Stokes et al., 2007; Martin‐Ventura et al., 2012), and ROS then further activate non‐adherent platelets and oxidized phospholipids including LDLs (oxLDL) (Freedman, 2008). OxLDL in turn stimulates platelet–monocyte aggregate formation, which promotes phenotypic changes in monocytes, monocyte extravasation and enhanced foam‐cell formation in vitro and in vivo. Inhibition of P2Y1 and P2Y12 receptors partly blocked oxLDL‐induced platelet–monocyte aggregation (Badrnya et al., 2014) (Figure 3).
Figure 3.
Effects of P2Y12 receptor activation on endothelial cell (EC), platelets and vascular smooth muscle cells (VSMC) function. For detailed explanation, see text. miR: micro RNA.
NO is an endogenous platelet inhibitor (Freedman, 2008), and platelet sensitivity towards NO is reduced in the presence of P2Y12 receptor activation (Kirkby et al., 2013). As increased ROS formation by platelets may reduce NO availability, this should indirectly increase platelet activation and vascular tone. Activation of P2Y12 receptors may occur early during atherosclerosis because circulating levels of the P2Y12 receptor agonist ADP were elevated in patients with peripheral atherosclerotic disease especially of younger age (Jalkanen et al., 2015). In patients with peripheral atherosclerotic disease, inhibition of P2Y12 receptors greatly increased platelet sensitivity towards NO (factor of 1000) thereby attenuating platelet activation and aggregation.
Studies using P2Y12 knockout mice and bone marrow chimeric mice expressing either vessel wall or platelet P2Y12 receptors indicate a contribution of the platelet P2Y12 receptor to restenosis (Evans et al., 2009) and of the vessel wall and platelet P2Y12 receptors to the development of atherosclerosis (Li et al., 2012; West et al., 2014).
Thienopyridines
In rabbits, with high‐fat diet‐induced atherosclerosis, treatment with clopidogrel attenuated plaque development and reduced the vascular wall and systemic inflammation (Li et al., 2007; Hadi et al., 2013). Two studies in ApoE knockout mice demonstrated beneficial effects of clopidogrel in attenuating atherosclerotic lesion formation (Afek et al., 2009; Takeda et al., 2012). However, other studies in ApoE knockout mice suggested that clopidogrel may not be effective in preventing early atherosclerosis (Schulz et al., 2008; West et al., 2014). Clopidogrel has also been shown to reduce neointimal formation (Waksman et al., 2008; Akbulut et al., 2004). Finally, an association between increased platelet reactivity during clopidogrel treatment and greater coronary artery atherosclerotic disease burden and plaque calcification has been described in patients undergoing PCI (Chirumamilla et al., 2012).
Ticagrelor
Supporting the role of platelets in the initiation of atherosclerosis, 8 weeks of ticagrelor treatment attenuated the initiation of atherosclerosis in hypercholesterolemic ApoE deficient mice (Schirmer et al., 2012). However, a similar study using the same model with only 4 weeks treatment showed no effect of ticagrelor or clopidogrel treatment on atherosclerosis development (West et al., 2014). The discrepancy between these studies can potentially be explained by drug dose, the time of initiation of treatment and the duration of the treatment.
Adenosine is a modulator of inflammatory responses and adenosine receptor deficient mice on an ApoE‐deficient background display alterations in atherosclerosis development (Jones et al., 2004; Reiss et al., 2004; Wang et al., 2009; Bingham et al., 2010; Koupenova et al., 2012; Teng et al., 2014) with more clear benefit seen in CD73 (ecto‐5′‐nucleotidase) and ApoE double knockout mice, which should have reduced adenosine production (Buchheiser et al., 2011). To address the potential impact of ticagrelor on established atherosclerosis, 20 weeks old ApoE knockout mice with established plaques were treated with ticagrelor for 25 weeks. In this study, ticagrelor induced a more stable plaque phenotype (thicker fibrous cap and reduced necrotic core) with a trend for a reduction in plaque size (Rusnak et al., 2014). Additionally, ticagrelor has been shown to reduce neointimal formation in a mouse carotid artery injury model (Patil et al., 2010) and reduced neointimal hyperplasia in a rabbit carotid anastomosis model (Sürer et al., 2014).
In summary, there is data from animal models supporting the hypothesis that thienopyridines and ticagrelor can affect the initiation and progression of atherosclerosis primarily via inhibition of P2Y12 receptors. Patient imaging studies are needed to validate the animal findings and to compare thienopyridines and ticagrelor with regards to the development and progression of atherosclerosis.
Cardioprotection
Thienopyridines
In rabbits, pre‐treatment with clopidogrel for 2 or 3 days (Yang et al., 2013) reduced infarct size following ischaemia/reperfusion. Interestingly, the protective effect of clopidogrel could be attenuated with MRS1754, an adenosine A2B receptor antagonist (Ye et al., 2013). When dosed to an equal P2Y12 receptor blockade, ticagrelor but not clopidogrel reduced infarct size in ischaemia/reperfusion models both in dogs and rats (further discussed below; Wang et al., 2010; Nanhwan et al., 2014; Ye et al., 2015). The explanation for the differences between the these studies concerning the cardioprotective effect of clopidogrel is unclear, but might be related to the species, the dose and treatment duration of clopidogrel or the length of reperfusion.
In patients, infarct size was significantly smaller (MRI and enzymic data) and myocardial salvage index was higher, in patients receiving clopidogrel 600 mg compared with 300 mg (Song et al., 2012). Whether this beneficial effect of clopidogrel resulted from direct effects on cardioprotective signalling cascades as suggested by (Yang et al. 2013) or was related to a more potent and quicker platelet inhibition with a 600 mg clopidogrel loading dose remains to be established. In support of the latter hypothesis, clopidogrel improved myocardial blush grade in the 600 mg group more than in the 300 mg group, supporting the notion of improved perfusion and less platelet aggregation (Song et al., 2012).
Ticagrelor
In a dog model of myocardial ischaemia/reperfusion, ticagrelor initiated 5 min before reperfusion reduced infarct size, whereas clopidogrel had no effect despite a similar and almost complete P2Y12 receptor inhibition for clopidogrel and ticagrelor (Wang et al., 2010). Thus, these data indicate a non‐P2Y12 receptor‐mediated cardioprotective effect of ticagrelor. In a rat ischaemia/reperfusion model, ticagrelor but not clopidogrel dose‐dependently reduced infarct size after 7 days p.o. pre‐treatment. The effect was reversed by adenosine receptor blockade providing evidence that the seen cardioprotective effect of ticagrelor was adenosine‐dependent (Nanhwan et al., 2014). In a recent follow‐up study using the same rat model, a single acute administration of ticagrelor just before reperfusion provided a similar reduction in infarct size following ischaemia/reperfusion as previously seen with p.o. pre‐treatment. Again, clopidogrel was ineffective. In addition, infarct size reduction translated into an improved heart function 4 weeks later. Finally, delayed initiation of p.o. ticagrelor treatment starting the day after reperfusion provided a similar improvement in heart function 4 weeks later as that following the single acute treatment (Ye et al., 2015). If these data translate into man, exposure of ticagrelor at the time of PCI could protect against reperfusion injury and improve long‐term heart function. Interestingly, the data indicate that an improved heart function may not exclusively depend on an acute cardioprotective effect as delayed exposure to ticagrelor provided a long‐term benefit. This may be especially relevant for patients with ST‐elevation myocardial infarction (STEMI) that have a delayed increase in the plasma concentration of ticagrelor and thienopyridines (Alexopoulos et al., 2012). In the first rat study, the infarct size reduction by ticagrelor was partly reversed by high‐dose, but not low‐dose, aspirin (Nanhwan et al., 2014) providing a plausible mechanistic explanation for the apparent interaction with high‐dose aspirin in patients receiving ticagrelor seen in the PLATO trial (Mahaffey et al., 2011). Previously, an alternative hypothesis has been proposed related to that the balance between the added anti‐platelet benefit of high‐dose aspirin in combination with P2Y12 receptor antagonism and its potential harm may be different depending on the level of P2Y12 receptor blockade (Kirkby et al., 2011; Björkman et al., 2013; see also Warner et al., 2011). These hypotheses do not contradict each other, but could work together to explain the apparent interaction.
The adenosine‐dependent cardioprotective effect of ticagrelor in animal models may help explain the unique cardiovascular and total mortality benefit seen with ticagrelor in the PLATO trial, but this remains to be shown in man. In the recent PEGASUS study, both 60 and 90 mg of ticagrelor did show a similar beneficial effect on the primary composite endpoint. There was a non‐significant trend for an effect on cardiovascular mortality on both 60 and 90 mg and a numerical decrease in total mortality on 60, but not 90 mg ticagrelor. As the study was not powered to show significant effects on cardiovascular or total mortality, these data need to be interpreted with caution.
The stronger and more rapid platelet inhibition, compared with that after clopidogrel, did not translate into a greater benefit for ticagrelor, in terms of myocardial perfusion in the PLATO angiographic sub‐study (Kunadian et al., 2013). Also, in the ATLANTIC study, when pre‐hospital and in‐hospital ticagrelor administration was compared, there was no difference in terms of myocardial perfusion (Montalescot et al., 2014). Potentially, the short time interval between randomization and PCI, in the PLATO angiographic sub‐study (median 0.68 h), and between pre‐hospital and in‐hospital administration in the ATLANTIC study (median 31 min) was too short to induce any significant difference in the early pharmacodynamic effect between treatments to impact on myocardial perfusion.
Randomized clinical studies comparing the effect of thienopyridines and ticagrelor in terms of infarct size following ischaemia/reperfusion as well as myocardial perfusion are needed. Two such studies are listed on ClinicalTrials.gov. (NCT02233790, NCT02429271) and the design of another has been published (Park et al., 2014). Given the issue of rapid drug absorption specifically in STEMI patients, highlighted previously, the design of such patient studies are challenging.
In summary, experimental evidence support the possibility that thienopyridines and ticagrelor can reduce infarct size following ischaemia/reperfusion. The evidence is especially convincing for ticagrelor as the underlying mechanism, mediated by adenosine, is defined and clear benefit over clopidogrel has been shown in comparative studies.
Conclusion
The effect and clinical benefit of P2Y12 receptor antagonists may not be limited to platelet inhibition and prevention of arterial thrombus formation. Potential additional effects include those secondary to platelet inhibition such as reducing the pro‐inflammatory role of platelets and those that may be related to P2Y12 receptor inhibition on other cells apart from platelets including dyspnea. The thienopyridines and ticagrelor differ in their mode of action on the P2Y12 receptor, and in addition, ticagrelor is unique in having the only well‐documented additional target for inhibition, the adenosine transporter ENT1. These differences provide different opportunities for the P2Y12 receptor antagonists to have effects beyond protection against arterial thrombosis. Explorative studies in humans are needed to translate promising animal data in terms of cardioprotection, vascular function, inflammation and atherosclerosis and to compare the effects of clopidogrel, prasugrel and ticagrelor.
Conflict of interest
Rainer Schulz: research grants from Zealand Pharma; honoraria for lectures and advisory boards from AstraZeneca, Recordati, Sanofi and Servier. Sven Nylander is an employee of AstraZeneca.
Nylander, S. , and Schulz, R. (2016) Effects of P2Y12 receptor antagonists beyond platelet inhibition – comparison of ticagrelor with thienopyridines. British Journal of Pharmacology, 173: 1163–1178. doi: 10.1111/bph.13429.
References
- Afek A, Kogan E, Maysel‐Auslender S, Mor A, Regev E, Rubinstein A, et al. (2009). Clopidogrel attenuates atheroma formation and induces a stable plaque phenotype in apolipoprotein E knockout mice. Microvasc Res 77: 364–369. [DOI] [PubMed] [Google Scholar]
- Akbulut M, Ozbay Y, Karaca I, Ilkay E, Gundogdu O, Arslan N (2004). The effect of long‐term clopidogrel use on neointimal formation after percutaneous coronary intervention. Coron Artery Dis 15: 347–352. [DOI] [PubMed] [Google Scholar]
- Aleil B, Ravanat C, Cazenave JP, Rochoux G, Heitz A, Gachet C (2005). Flow cytometric analysis of intraplatelet VASP phosphorylation for the detection of clopidogrel resistance in patients with ischemic cardiovascular diseases. J Thromb Haemost 3: 85–92. [DOI] [PubMed] [Google Scholar]
- Alexander SPH, Davenport AP, Kelly E, Marrion N, Peters JA, Benson HE, et al. (2015a). The Concise Guide to PHARMACOLOGY 2015/16: G Protein‐Coupled Receptors. Br J Pharmacol 172: 5744–5869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Fabbro D, Kelly E, Marrion N, Peters JA, Benson HE, et al. (2015b). The Concise Guide to PHARMACOLOGY 2015/16: Enzymes. Br J Pharmacol 172: 6024–6109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexopoulos D, Xanthopoulou I, Gkizas V, Kassimis G, Theodoropoulos KC, Makris G, et al. (2012). Randomized assessment of ticagrelor versus prasugrel antiplatelet effects in patients with ST‐segment‐elevation myocardial infarction. Circ Cardiovasc Interv 5: 797–804. [DOI] [PubMed] [Google Scholar]
- Alexopoulos D, Moulias A, Koutsogiannis N, Xanthopoulou I, Kakkavas A, Mavronasiou E, et al. (2013). Differential effect of ticagrelor versus prasugrel on coronary blood flow velocity in patients with non‐ST‐elevation acute coronary syndrome undergoing percutaneous coronary intervention: an exploratory study. Circ Cardiovasc Interv 6: 277–283. [DOI] [PubMed] [Google Scholar]
- Alsharif KF, Thomas MR, Judge HM, Khan H, Prince LR, Sabroe I, et al. (2015). Ticagrelor potentiates adenosine‐induced stimulation of neutrophil chemotaxis and phagocytosis. Vascul Pharmacol 71: 201–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Angiolillo DJ, Fernandez‐Ortiz A, Bernardo E, Ramírez C, Sabaté M, Jimenez‐Quevedo P, et al. (2006). Clopidogrel withdrawal is associated with proinflammatory and prothrombotic effects in patients with diabetes and coronary artery disease. Diabetes 55: 780–784. [DOI] [PubMed] [Google Scholar]
- Antonino MJ, Mahla E, Bliden KP, Tantry US, Gurbel PA (2009). Effect of long‐term clopidogrel treatment on platelet function and inflammation in patients undergoing coronary arterial stenting. Am J Cardiol 103: 1546–1550. [DOI] [PubMed] [Google Scholar]
- Armstrong D, Summers C, Ewart L, Nylander S, Sidaway JE, van Giezen JJ (2014). Characterization of the adenosine pharmacology of ticagrelor reveals therapeutically relevant inhibition of equilibrative nucleoside transporter 1. J Cardiovasc Pharmacol Ther 19: 209–219. [DOI] [PubMed] [Google Scholar]
- Aslam M, Schluter KD, Rohrbach S, Rafiq A, Nazli S, Piper HM, et al. (2013). Hypoxia‐reoxygenation‐induced endothelial barrier failure: role of RhoA, Rac1 and myosin light chain kinase. J Physiol 591: 461–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aslam M, Tanislav C, Troidl C, Schulz R, Hamm C, Gündüz D (2014). cAMP controls the restoration of endothelial barrier function after thrombin‐induced hyperpermeability via Rac1 activation. Physiol Rep 2 pii: e12175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Azar RR, Kassab R, Zoghbi A, Aboujaoudé S, El‐Osta H, Ghorra P, et al. (2006). Effects of clopidogrel on soluble CD40 ligand and on high‐sensitivity C‐reactive protein in patients with stable coronary artery disease. Am Heart J 151: e1–521. [DOI] [PubMed] [Google Scholar]
- Baaten CC, Veenstra LF, Wetzels R, van Geffen JP, Swieringa F, de Witt SM, et al. (2015). Gradual increase in thrombogenicity of juvenile platelets formed upon offset of prasugrel medication. Haematologica 100: 1131–1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Badrnya S, Schrottmaier WC, Kral JB, Yaiw KC, Volf I, Schabbauer G, et al. (2014). Platelets mediate oxidized low‐density lipoprotein‐induced monocyte extravasation and foam cell formation. Arterioscler Thromb Vasc Biol 34: 571–580. [DOI] [PubMed] [Google Scholar]
- Ben Addi A, Cammarata D, Conley PB, Boeynaems JM, Robaye B (2010). Role of the P2Y12 receptor in the modulation of murine dendritic cell function by ADP. J Immunol 185: 5900–5906. [DOI] [PubMed] [Google Scholar]
- Bernlochner I, Goedel A, Plischke C, Schüpke S, Haller B, Schulz C, et al. (2015). Impact of immature platelets on platelet response to ticagrelor and prasugrel in patients with acute coronary syndrome. Eur Heart J 36: 3202–3210. [DOI] [PubMed] [Google Scholar]
- Bhatt DL, Lincoff AM, Gibson CM, Stone GW, McNulty S, Montalescot G, et al. (2009). Intravenous platelet blockade with cangrelor during PCI. N Engl J Med 361: 2330–2341. [DOI] [PubMed] [Google Scholar]
- Bhatt DL, Stone GW, Mahaffey KW, Gibson CM, Steg PG, Hamm CW, et al. (2013). Effect of platelet inhibition with cangrelor during PCI on ischemic events. N Engl J Med 368: 1303–1313. [DOI] [PubMed] [Google Scholar]
- Bingham TC, Fisher EA, Parathath S, Reiss AB, Chan ES, Cronstein BN (2010). A2A adenosine receptor stimulation decreases foam cell formation by enhancing ABCA1‐dependent cholesterol efflux. J Leukoc Biol 87: 683–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Björkman JA, Zachrisson H, Forsberg GB, von Bahr H, Hansson GI, Warner TD, et al. (2013). High‐dose aspirin in dogs increases vascular resistance with limited additional anti‐platelet effect when combined with potent P2Y12 inhibition. Thromb Res 131: 313–319. [DOI] [PubMed] [Google Scholar]
- Bonaca MP, Bhatt DL, Cohen M, Steg PG, Storey RF, Jensen EC, et al. (2015). Long‐Term use of ticagrelor in patients with prior myocardial infarction. N Engl J Med 372: 1791–1800. [DOI] [PubMed] [Google Scholar]
- Bonello L, Harhouri K, Sabatier F, Camoin‐Jau L, Arnaud L, Baumstarck‐Barrau K, et al. (2010). Level of adenosine diphosphate receptor P2Y12 blockade during percutaneous coronary intervention predicts the extent of endothelial injury, assessed by circulating endothelial cell measurement. J Am Coll Cardiol 56: 1024–1031. [DOI] [PubMed] [Google Scholar]
- Bonello L, Laine M, Kipson N, Mancini J, Helal O, Fromonot J, et al. (2013). Ticagrelor increases adenosine plasma concentration in patients with an acute coronary syndrome. J Am Coll Cardiol 63: 872–877. [DOI] [PubMed] [Google Scholar]
- Bonello L, Frere C, Cointe S, Laine M, Mancini J, Thuny F, et al. (2015). Ticagrelor increases endothelial progenitor cell level compared to clopidogrel in acute coronary syndromes: A prospective randomized study. Int J Cardiol 187: 502–507. [DOI] [PubMed] [Google Scholar]
- Braun OO, Johnell M, Varenhorst C, James S, Brandt JT, Jakubowski JA, et al. (2008). Greater reduction of platelet activation markers and platelet‐monocyte aggregates by prasugrel compared to clopidogrel in stable coronary artery disease. Thromb Haemost 100: 626–633. [PubMed] [Google Scholar]
- Buchheiser A, Ebner A, Burghoff S, Ding Z, Romio M, Viethen C, et al. (2011). Inactivation of CD73 promotes atherogenesis in apolipoprotein E‐deficient mice. Cardiovasc Res 92: 338–347. [DOI] [PubMed] [Google Scholar]
- Burki NK, Dale WJ, Lee LY (2005). Intravenous adenosine and dyspnea in humans. J Appl Physiol 98: 180–185. [DOI] [PubMed] [Google Scholar]
- Cattaneo M, Faioni EM (2012). Why does ticagrelor induce dyspnea? Thromb Haemost 108: 1031–1036. [DOI] [PubMed] [Google Scholar]
- Cattaneo M, Schulz R, Nylander S (2014). Adenosine‐mediated effects of ticagrelor: evidence and potential clinical relevance. J Am Coll Cardiol 63: 2503–2509. [DOI] [PubMed] [Google Scholar]
- Cesari F, Marcucci R, Gori AM, Caporale R, Fanelli A, Casola G, et al. (2013). Reticulated platelets predict cardiovascular death in acute coronary syndrome patients. Insights from the AMI‐Florence 2 Study. Thromb Haemost 109: 846–853. [DOI] [PubMed] [Google Scholar]
- Chen YG, Xu F, Zhang Y, Ji QS, Sun Y, Lü RJ, et al. (2006). Effect of aspirin plus clopidogrel on inflammatory markers in patients with non‐ST‐segment elevation acute coronary syndrome. Chin Med J (Engl) 119: 32–36. [PubMed] [Google Scholar]
- Chirumamilla AP, Maehara A, Mintz GS, Mehran R, Kanwal S, Weisz G, et al. (2012). High platelet reactivity on clopidogrel therapy correlates with increased coronary atherosclerosis and calcification: a volumetric intravascular ultrasound study. JACC Cardiovasc Imaging 5: 540–549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diehl P, Olivier C, Halscheid C, Helbing T, Bode C, Moser M (2010). Clopidogrel affects leukocyte dependent platelet aggregation by P2Y12 expressing leukocytes. Basic Res Cardiol 105: 379–387. [DOI] [PubMed] [Google Scholar]
- Dong ZM, Brown AA, Wagner DD (2000). Prominent role of P‐selectin in the development of advanced atherosclerosis in ApoE‐deficient mice. Circulation 101: 2290–2295. [DOI] [PubMed] [Google Scholar]
- Evangelista V, Manarini S, Dell'Elba G, Martelli N, Napoleone E, Di Santo A, et al. (2005). Clopidogrel inhibits platelet‐leukocyte adhesion and platelet‐dependent leukocyte activation. Thromb Haemost 94: 568–577. [PubMed] [Google Scholar]
- Evans DJ, Jackman LE, Chamberlain J, Crosdale DJ, Judge HM, Jetha K, et al. (2009). Platelet P2Y(12) receptor influences the vessel wall response to arterial injury and thrombosis. Circulation 119: 116–122. [DOI] [PubMed] [Google Scholar]
- Freedman JE (2008). Oxidative stress and platelets. Arterioscler Thromb Vasc Biol 28: s11–s16. [DOI] [PubMed] [Google Scholar]
- Frelinger AL 3rd, Jakubowski JA, Li Y, Barnard MR, Fox ML, Linden MD, et al. (2007). The active metabolite of prasugrel inhibits ADP‐stimulated thrombo‐inflammatory markers of platelet activation: Influence of other blood cells, calcium, and aspirin. Thromb Haemost 98: 192–200. [PubMed] [Google Scholar]
- Froldi G, Bertin R, Dorigo P, Montopoli M, Caparrotta L (2011). Endothelium‐independent vasorelaxation by ticlopidine and clopidogrel in rat caudal artery. J Pharm Pharmacol 63: 1056–1062. [DOI] [PubMed] [Google Scholar]
- Fuentes E, Palomo I, Alarcón M (2015). Platelet miRNAs and cardiovascular diseases. Life Sci 133: 29–44. [DOI] [PubMed] [Google Scholar]
- Gachet C (2012). P2Y(12) receptors in platelets and other hematopoietic and non‐hematopoietic cells. Purinergic Signal 8: 609–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge H, Zhou Y, Liu X, Nie X, Wang Z, Guo Y, et al. (2012). Relationship between plasma inflammatory markers and platelet aggregation in patients with clopidogrel resistance after angioplasty. Angiology 63: 62–66. [DOI] [PubMed] [Google Scholar]
- Gent M, Beaumont D, Blanchard J, Bousser MG, Coffman J, Easton JD, et al. (1996). A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet 348: 1329–1339. [DOI] [PubMed] [Google Scholar]
- Giachini FR, Leite R, Osmond DA, Lima VV, Inscho EW, Webb RC, et al. (2014). Anti‐platelet therapy with clopidogrel prevents endothelial dysfunction and vascular remodeling in aortas from hypertensive rats. PLoS One 9: e91890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gremmel T, Eslam RB, Koppensteiner R, Lang IM, Panzer S (2013). Prasugrel reduces agonists' inducible platelet activation and leukocyte‐platelet interaction more efficiently than clopidogrel. Cardiovasc Ther 31: e40–e45. [DOI] [PubMed] [Google Scholar]
- Gros A, Ollivier V, Ho‐Tin‐Noé B (2015). Platelets in inflammation: regulation of leukocyte activities and vascular repair. Front Immunol 5: 678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grzesk G, Kozinski M, Navarese EP, Krzyzanowski M, Grzesk E, Kubica A, et al. (2012). Ticagrelor, but not clopidogrel and prasugrel, prevents ADP‐induced vascular smooth muscle cell contraction: a placebo‐controlled study in rats. Thromb Res 130: 65–69. [DOI] [PubMed] [Google Scholar]
- Gurbel PA, Bliden KP, Tantry US (2006). Effect of clopidogrel with and without eptifibatide on tumor necrosis factor‐alpha and C‐reactive protein release after elective stenting: results from the CLEAR PLATELETS 1b study. J Am Coll Cardiol 48: 2186–2191. [DOI] [PubMed] [Google Scholar]
- Gurbel PA, Tantry US (2015). Inhibited and Uninhibited Platelet Deposition Within a Thrombus: Does It Depend on the Antiplatelet Drug? Arterioscler Thromb Vasc Biol 35: 2081–2082. [DOI] [PubMed] [Google Scholar]
- Hadi NR, Mohammad BI, Ajeena IM, Sahib HH (2013). Antiatherosclerotic potential of clopidogrel: antioxidant and anti‐inflammatory approaches. Biomed Res Int 2013: 790263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamilos M, Muller O, Ntalianis A, Trana C, Bartunek J, Sarno G, et al. (2011). Relationship between peripheral arterial reactive hyperemia and residual platelet reactivity after 600 mg clopidogrel. J Thromb Thrombolysis 32: 64–71. [DOI] [PubMed] [Google Scholar]
- Harrington RA, Stone GW, McNulty S, White HD, Lincoff AM, Gibson CM, et al. (2009). Platelet inhibition with cangrelor in patients undergoing PCI. N Engl J Med 361: 2318–2329. [DOI] [PubMed] [Google Scholar]
- Heitzer T, Rudolph V, Schwedhelm E, Karstens M, Sydow K, Ortak M, et al. (2006). Clopidogrel improves systemic endothelial nitric oxide bioavailability in patients with coronary artery disease: evidence for antioxidant and antiinflammatory effects. Arterioscler Thromb Vasc Biol 26: 1648–1652. [DOI] [PubMed] [Google Scholar]
- Held C, Asenblad N, Bassand JP, Becker RC, Cannon CP, Claeys MJ, et al. (2011). Ticagrelor versus clopidogrel in patients with acute coronary syndromes undergoing coronary artery bypass surgery: results from the PLATO (Platelet Inhibition and Patient Outcomes) trial. J Am Coll Cardiol 57: 672–684. [DOI] [PubMed] [Google Scholar]
- Hoefer T, Armstrong PC, Finsterbusch M, Chan MV, Kirkby NS, Warner TD (2015). Drug‐Free Platelets Can Act as Seeds for Aggregate Formation During Antiplatelet Therapy. Arterioscler Thromb Vasc Biol 35: 2122–2133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hollopeter G, Jantzen HM, Vincent D, Li G, England L, Ramakrishnan V, et al. (2001). Identification of the platelet ADP receptor targeted by antithrombotic drugs. Nature 409: 202–207. [DOI] [PubMed] [Google Scholar]
- Högberg C, Svensson H, Gustafsson R, Eyjolfsson A, Erlinge D (2010). The reversible oral P2Y12 antagonist AZD6140 inhibits ADP‐induced contractions in murine and human vasculature. Int J Cardiol 142: 187–192. [DOI] [PubMed] [Google Scholar]
- Hulsmans M, Holvoet P (2013). MicroRNA‐containing microvesicles regulating inflammation in association with atherosclerotic disease. Cardiovasc Res 100: 7–18. [DOI] [PubMed] [Google Scholar]
- Husted S, Storey RF, Harrington RA, Emanuelsson H, Cannon CP (2010). Changes in inflammatory biomarkers in patients treated with ticagrelor or clopidogrel. Clin Cardiol 33: 206–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Husted SE, Storey RF, Bliden K, Tantry US, Høimark L, Butler K, et al. (2012). Pharmacokinetics and pharmacodynamics of ticagrelor in patients with stable coronary artery disease: results from the ONSET‐OFFSET and RESPOND studies. Clin Pharmacokinet 51: 397–409. [DOI] [PubMed] [Google Scholar]
- Ibrahim H, Schutt RC, Hannawi B, DeLao T, Barker CM, Kleiman NS (2014). Association of immature platelets with adverse cardiovascular outcomes. J Am Coll Cardiol 64: 2122–2129. [DOI] [PubMed] [Google Scholar]
- Ibrahim H, Nadipalli S, DeLao T, Guthikonda S, Kleiman NS (2012). Immature platelet fraction (IPF) determined with an automated method predicts clopidogrel hyporesponsiveness. J Thromb Thrombolysis 33: 137–142. [DOI] [PubMed] [Google Scholar]
- Jakubowski A, Chlopicki S, Olszanecki R, Jawien J, Lomnicka M, Dupin JP, et al. (2005). Endothelial action of thienopyridines and thienopyrimidinones in the isolated guinea pig heart. Prostaglandins Leukot Essent Fatty Acids 72: 139–145. [DOI] [PubMed] [Google Scholar]
- Jakubowski JA, Zhou C, Winters KJ, Lachno DR, Howard J, Payne CD, et al. (2014). The effect of prasugrel on ADP‐stimulated markers of platelet activation in patients with sickle cell disease. Platelets 20: 1–6. [DOI] [PubMed] [Google Scholar]
- Jalkanen J, Yegutkin GG, Hollmén M, Aalto K, Kiviniemi T, Salomaa V, et al. (2015). Aberrant circulating levels of purinergic signaling markers are associated with several key aspects of peripheral atherosclerosis and thrombosis. Circ Res 116: 1206–1215. [DOI] [PubMed] [Google Scholar]
- Järemo P, Lindahl TL, Fransson SG, Richter A (2002). Individual variations of platelet inhibition after loading doses of clopidogrel. J Intern Med 252: 233–238. [DOI] [PubMed] [Google Scholar]
- Johnston LR, La Flamme AC, Larsen PD, Harding SA (2015). Prasugrel inhibits platelet‐enhanced pro‐inflammatory CD4+ T cell responses in humans. Atherosclerosis 239: 283–286. [DOI] [PubMed] [Google Scholar]
- Jones MR, Zhao Z, Sullivan CP, Schreiber BM, Stone PJ, Toselli PA, et al. (2004). A(3) adenosine receptor deficiency does not influence atherogenesis. J Cell Biochem 92: 1034–1043. [DOI] [PubMed] [Google Scholar]
- Judge HM, Buckland RJ, Sugidachi A, Jakubowski JA, Storey RF (2008). The active metabolite of prasugrel effectively blocks the platelet P2Y12 receptor and inhibits procoagulant and pro‐inflammatory platelet responses. Platelets 19: 125–133. [DOI] [PubMed] [Google Scholar]
- Kawaguchi A, Sato M, Kimura M, Ichinohe T, Tazaki M, Shibukawa Y (2015). Expression and function of purinergic P2Y12 receptors in rat trigeminal ganglion neurons. Neurosci Res 98: 17–27. [DOI] [PubMed] [Google Scholar]
- Kirkby NS, Leadbeater PD, Chan MV, Nylander S, Mitchell JA, Warner TD (2011). Antiplatelet effects of aspirin vary with level of P2Y₁₂ receptor blockade supplied by either ticagrelor or prasugrel. J Thromb Haemost 9: 2103–2105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kirkby NS, Lundberg MH, Chan MV, Vojnovic I, Solomon AB, Emerson M, et al. (2013). Blockade of the purinergic P2Y12 receptor greatly increases the platelet inhibitory actions of nitric oxide. Proc Natl Acad Sci U S A 110: 15782–15787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koupenova M, Johnston‐Cox H, Vezeridis A, Gavras H, Yang D, Zannis V, et al. (2012). A2b adenosine receptor regulates hyperlipidemia and atherosclerosis. Circulation 125: 354–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuijpers MJ, Megens RT, Nikookhesal E, Feijge MA, De Mey JG, oude Egbrink MG, et al. (2011). Role of newly formed platelets in thrombus formation in rat after clopidogrel treatment: comparison to the reversible binding P2Y₁₂ antagonist ticagrelor. Thromb Haemost 106: 1179–1188. [DOI] [PubMed] [Google Scholar]
- Kunadian V, James SK, Wojdyla DM, Zorkun C, Wu J, Storey RF, et al. (2013). Angiographic outcomes in the PLATO Trial (Platelet Inhibition and Patient Outcomes). JACC Cardiovasc Interv 6: 671–683. [DOI] [PubMed] [Google Scholar]
- Lhermusier T, Baker NC, Waksman R (2015). Overview of the 2014 Food and Drug Administration Cardiovascular and Renal Drugs Advisory Committee meeting regarding cangrelor. Am J Cardiol 115: 1154–1161. [DOI] [PubMed] [Google Scholar]
- Li M, Zhang Y, Ren H, Zhang Y, Zhu X (2007). Effect of clopidogrel on the inflammatory progression of early atherosclerosis in rabbits model. Atherosclerosis 194: 348–356. [DOI] [PubMed] [Google Scholar]
- Li D, Wang Y, Zhang L, Luo X, Li J, Chen X, et al. (2012). Roles of purinergic receptor P2Y, G protein‐coupled 12 in the development of atherosclerosis in apolipoprotein E‐deficient mice. Arterioscler Thromb Vasc Biol 32: e81–e89. [DOI] [PubMed] [Google Scholar]
- Lindholm D, Storey RF, Christersson C, Halvorsen S, Grove EL, Braun OÖ, et al. (2015). Design and rationale of TROCADERO: A TRial Of Caffeine to Alleviate DyspnEa Related to ticagrelOr. Am Heart J 170: 465–470. [DOI] [PubMed] [Google Scholar]
- Liverani E, Rico MC, Garcia AE, Kilpatrick LE, Kunapuli SP (2013). Prasugrel metabolites inhibit neutrophil functions. J Pharmacol Exp Ther 344: 231–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lum H, Jaffe HA, Schulz IT, Masood A, RayChaudhury A, Green RD (1999). Expression of PKA inhibitor (PKI) gene abolishes cAMP‐mediated protection to endothelial barrier dysfunction. Am J Physiol 277: C580–C588. [DOI] [PubMed] [Google Scholar]
- Lussana F, Di Marco F, Terraneo S, Parati M, Razzari C, Scavone M, et al. (2015). Effect of prasugrel in patients with asthma: results of PRINA, a randomized, double‐blind, placebo‐controlled, cross‐over study. J Thromb Haemost 13: 136–141. [DOI] [PubMed] [Google Scholar]
- Mahaffey KW, Wojdyla DM, Carroll K, Becker RC, Storey RF, Angiolillo DJ, et al. (2011). Ticagrelor compared with clopidogrel by geographic region in the Platelet Inhibition and Patient Outcomes (PLATO) trial. Circulation 124: 544–554. [DOI] [PubMed] [Google Scholar]
- Malek LA, Grabowski M, Spiewak M, Filipiak KJ, Szpotanska M, Imiela T, et al. (2007). Relation between impaired antiplatelet response to clopidogrel and possible pleiotropic effects. J Thromb Thrombolysis 24: 301–305. [DOI] [PubMed] [Google Scholar]
- Martin‐Ventura JL, Madrigal‐Matute J, Martinez‐Pinna R, Ramos‐Mozo P, Blanco‐Colio LM, Moreno JA, et al. (2012). Erythrocytes, leukocytes and platelets as a source of oxidative stress in chronic vascular diseases: detoxifying mechanisms and potential therapeutic options. Thromb Haemost 108: 435–442. [DOI] [PubMed] [Google Scholar]
- Massberg S, Brand K, Grüner S, Page S, Müller E, Müller I, et al. (2002). A critical role of platelet adhesion in the initiation of atherosclerotic lesion formation. J Exp Med 196: 887–896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molero L, López‐Farré A, Mateos‐Cácere PJ, Fernández‐Sánchez R, Luisa Maestro M, Silva J, et al. (2005). Effect of clopidogrel on the expression of inflammatory markers in rabbit ischemic coronary artery. Br J Pharmacol 146: 419–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Montalescot G, Sideris G, Meuleman C, Baldit‐Sollier C, Lellouche N, Steg PG, et al. (2006). A randomized comparison of high clopidogrel loading doses in patients with non‐ST‐segment elevation acute coronary syndromes: the ALBION (Assessment of the Best Loading Dose of Clopidogrel to Blunt Platelet Activation, Inflammation and Ongoing Necrosis) trial. J Am Coll Cardiol 48: 931–938. [DOI] [PubMed] [Google Scholar]
- Montalescot G, van ‘t Hof AW, Lapostolle F, Silvain J, Lassen JF, Bolognese L, et al. ATLANTIC Investigators(2014). Prehospital ticagrelor in ST‐segment elevation myocardial infarction. N Engl J Med 371: 1016–1027. [DOI] [PubMed] [Google Scholar]
- Muhlestein JB (2010). Effect of antiplatelet therapy on inflammatory markers in atherothrombotic patients. Thromb Haemost 103: 71–82. [DOI] [PubMed] [Google Scholar]
- Nanhwan MK, Ling S, Kodakandla M, Nylander S, Ye Y, Birnbaum Y (2014). Chronic treatment with ticagrelor limits myocardial infarct size: an adenosine and cyclooxygenase‐2‐dependent effect. Arterioscler Thromb Vasc Biol 34: 2078–2085. [DOI] [PubMed] [Google Scholar]
- Nording HM, Seizer P, Langer HF (2015). Platelets in inflammation and atherogenesis. Front Immunol 6: 98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nylander S, Femia EA, Scavone M, Berntsson P, Asztély AK, Nelander K, et al. (2013). Ticagrelor inhibits human platelet aggregation via adenosine in addition to P2Y12 antagonism. J Thromb Haemost 11: 1867–1876. [DOI] [PubMed] [Google Scholar]
- Öhman J, Kudira R, Albinsson S, Olde B, Erlinge D (2012). Ticagrelor induces adenosine triphosphate release from human red blood cells. Biochem Biophys Res Commun 418: 754–758. [DOI] [PubMed] [Google Scholar]
- Ostad MA, Nick E, Paixao‐Gatinho V, Schnorbus B, Schiewe R, Tschentscher P, et al. (2011). Lack of evidence for pleiotropic effects of clopidogrel on endothelial function and inflammation in patients with stable coronary artery disease: results of the double‐blind, randomized CASSANDRA study. Clin Res Cardiol 100: 29–36. [DOI] [PubMed] [Google Scholar]
- Palmerini T, Barozzi C, Tomasi L, Sangiorgi D, Marzocchi A, De Servi S, et al. (2010). A randomised study comparing the antiplatelet and antiinflammatory effect of clopidogrel 150 mg/day versus 75 mg/day in patients with ST‐segment elevation acute myocardial infarction and poor responsiveness to clopidogrel: results from the DOUBLE study. Thromb Res 125: 309–314. [DOI] [PubMed] [Google Scholar]
- Park SD, Baek YS, Woo SI, Kim SH, Shin SH, Kim DH, et al. (2014). Comparing the effect of clopidogrel versus ticagrelor on coronary microvascular dysfunction in acute coronary syndrome patients (TIME trial): study protocol for a randomized controlled trial. Trials 15: 151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patil SB, Jackman LE, Francis SE, Judge HM, Nylander S, Storey RF (2010). Ticagrelor effectively and reversibly blocks murine platelet P2Y12‐mediated thrombosis and demonstrates a requirement for sustained P2Y12 inhibition to prevent subsequent neointima. Arterioscler Thromb Vasc Biol 30: 2385–2391. [DOI] [PubMed] [Google Scholar]
- Patti G, Grieco D, Dicuonzo G, Pasceri V, Nusca A, Di Sciascio G (2011). High versus standard clopidogrel maintenance dose after percutaneous coronary intervention and effects on platelet inhibition, endothelial function, and inflammation results of the ARMYDA‐150 mg (antiplatelet therapy for reduction of myocardial damage during angioplasty) randomized study. J Am Coll Cardiol 57: 771–778. [DOI] [PubMed] [Google Scholar]
- Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SPH, Buneman OP, et al. NC‐IUPHAR(2014). The IUPHAR/BPS Guide to PHARMACOLOGY: an expert‐driven knowledge base of drug targets and their ligands. Nucl. Acids Res. 42 (Database Issue): D1098–D1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quinn MJ, Bhatt DL, Zidar F, Vivekananthan D, Chew DP, Ellis SG, et al. (2004). Effect of clopidogrel pretreatment on inflammatory marker expression in patients undergoing percutaneous coronary intervention. Am J Cardiol 93: 679–684. [DOI] [PubMed] [Google Scholar]
- Rahman M, Gustafsson D, Wang Y, Thorlacius H, Braun OO (2014). Ticagrelor reduces neutrophil recruitment and lung damage in abdominal sepsis. Platelets 25: 257–263. [DOI] [PubMed] [Google Scholar]
- Ramadan R, Dhawan SS, Syed H, Pohlel FK, Binongo JN, Ghazzal ZB, et al. (2014). Effects of clopidogrel therapy on oxidative stress, inflammation, vascular function, and progenitor cells in stable coronary artery disease. J Cardiovasc Pharmacol 63: 369–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rauch BH, Rosenkranz AC, Ermler S, Böhm A, Driessen J, Fischer JW, et al. (2010). Regulation of functionally active P2Y12 ADP receptors by thrombin in human smooth muscle cells and the presence of P2Y12 in carotid artery lesions. Arterioscler Thromb Vasc Biol 30: 2434–2442. [DOI] [PubMed] [Google Scholar]
- Reiner MF, Stivala S, Akhmedov A, Spescha RD, Camici G, Beer JHJ (2013). Ticagrelor and endothelial dysfunction: platelet independent effects. J Thromb Haemost 11 (S2) Meeting abstract PA 1.01‐6. [Google Scholar]
- Reiss AB, Rahman MM, Chan ES, Montesinos MC, Awadallah NW, Cronstein BN (2004). Adenosine A2A receptor occupancy stimulates expression of proteins involved in reverse cholesterol transport and inhibits foam cell formation in macrophages. J Leukoc Biol 76: 727–734. [DOI] [PubMed] [Google Scholar]
- Rosiak M, Postula M, Kaplon‐Cieslicka A, Kondracka A, Trzepla E, Czlonkowski A, et al. (2013). Effect of ASA dose doubling versus switching to clopidogrel on plasma inflammatory markers concentration in patients with type 2 diabetes and high platelet reactivity: the AVOCADO study. Cardiol J 20: 545–551. [DOI] [PubMed] [Google Scholar]
- Rusnak J, Mogler C, Buttler A, Bea F, Blessing E, Katus HA, et al. (2014). Ticagrelor promotes atherosclerotic lesion stabilization in advanced lesions of Apolipoprotein E deficient mice. Clin Res Cardiol 103 (S1) Abstract. [Google Scholar]
- Satonaka H, Nagata D, Takahashi M, Kiyosue A, Myojo M, Fujita D, et al. (2015). Involvement of P2Y12 receptor in vascular smooth muscle inflammatory changes via MCP‐1 upregulation and monocyte adhesion. Am J Physiol Heart Circ Physiol 308: H853–H861. [DOI] [PubMed] [Google Scholar]
- Schäfer A, Fraccarollo D, Pförtsch S, Loch E, Neuser J, Vogt C, et al. (2011). Clopidogrel improves endothelial function and NO bioavailability by sensitizing adenylyl cyclase in rats with congestive heart failure. Basic Res Cardiol 106: 485–494. [DOI] [PubMed] [Google Scholar]
- Schirmer SH, Kratz MT, Kazakov A, Nylander S, Baumhakel M, Laufs U, et al. (2012). Inhibition of the adenosine diphosphate receptor P2Y12 reduces atherosclerotic plaque size in hypercholesterolemic ApoE−/− mice. Eur Heart J 33 (Supplement): A200. [Google Scholar]
- Schnorbus B, Daiber A, Jurk K, Warnke S, König J, Krahn U, et al. (2014). Effects of clopidogrel, prasugrel and ticagrelor on endothelial function, inflammatory and oxidative stress parameters and platelet function in patients undergoing coronary artery stenting for an acute coronary syndrome. A randomised, prospective, controlled study. BMJ Open 4: e005268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schrottmaier WC, Kral JB, Badrnya S, Assinger A (2015). Aspirin and P2Y12 Inhibitors in platelet‐mediated activation of neutrophils and monocytes. Thromb Haemost 114: 478–89. [DOI] [PubMed] [Google Scholar]
- Schulz C, Konrad I, Sauer S, Orschiedt L, Koellnberger M, Lorenz R, et al. (2008). Effect of chronic treatment with acetylsalicylic acid and clopidogrel on atheroprogression and atherothrombosis in ApoE‐deficient mice in vivo. Thromb Haemost 99: 190–195. [DOI] [PubMed] [Google Scholar]
- Shanker G, Kontos JL, Eckman DM, Wesley‐Farrington D, Sane DC (2006). Nicotine upregulates the expression of P2Y12 on vascular cells and megakaryoblasts. J Thromb Thrombolysis 22: 213–220. [DOI] [PubMed] [Google Scholar]
- Siasos G, Oikonomou E, Zaromitidou M, Kioufis S, Kokkou E, Mourouzis K, et al. (2015). Clopidogrel response variability is associated with endothelial dysfunction in coronary artery disease patients receiving dual antiplatelet therapy. Atherosclerosis 242: 102–108. [DOI] [PubMed] [Google Scholar]
- Simon J, Filippov AK, Göransson S, Wong YH, Frelin C, Michel AD, et al. (2002). Characterization and channel coupling of the P2Y(12) nucleotide receptor of brain capillary endothelial cells. J Biol Chem 277: 31390–31400. [DOI] [PubMed] [Google Scholar]
- Smadja DM, Bura A, Szymezak J, Blanchard A, Remones V, Azizi M, et al. (2012). Effect of clopidogrel on circulating biomarkers of angiogenesis and endothelial activation. J Cardiol 59: 30–35. [DOI] [PubMed] [Google Scholar]
- Solheim S, Pettersen AA, Arnesen H, Seljeflot I (2006). No difference in the effects of clopidogrel and aspirin on inflammatory markers in patients with coronary heart disease. Thromb Haemost 96: 660–664. [PubMed] [Google Scholar]
- Song YB, Hahn JY, Gwon HC, Chang SA, Lee SC, Choe YH, et al. (2012). A high loading dose of clopidogrel reduces myocardial infarct size in patients undergoing primary percutaneous coronary intervention: a magnetic resonance imaging study. Am Heart J 163: 500–507. [DOI] [PubMed] [Google Scholar]
- Springthorpe B, Bailey A, Barton P, Birkinshaw TN, Bonnert RV, Brown RC, et al. (2007). From ATP to AZD6140: the discovery of an orally active reversible P2Y12 receptor antagonist for the prevention of thrombosis. Bioorg Med Chem Lett 17: 6013–6018. [DOI] [PubMed] [Google Scholar]
- Steinhubl SR, Badimon JJ, Bhatt DL, Herbert JM, Lüscher TF (2007). Clinical evidence for anti‐inflammatory effects of antiplatelet therapy in patients with atherothrombotic disease. Vasc Med 12: 113–122. [DOI] [PubMed] [Google Scholar]
- Stokes KY, Russell JM, Jennings MH, Alexander JS, Granger DN (2007). Platelet‐associated NAD(P)H oxidase contributes to the thrombogenic phenotype induced by hypercholesterolemia. Free Radic Biol Med 43: 22–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Storey RF, Becker RC, Harrington RA, Husted S, James SK, Cools F, et al. (2011). Characterization of dyspnoea in PLATO study patients treated with ticagrelor or clopidogrel and its association with clinical outcomes. Eur Heart J 32: 2945–2953. [DOI] [PubMed] [Google Scholar]
- Storey RF, Husted S, Harrington RA, Heptinstall S, Wilcox RG, Peters G, et al. (2007). Inhibition of platelet aggregation by AZD6140, a reversible oral P2Y12 receptor antagonist, compared with clopidogrel in patients with acute coronary syndromes. J Am Coll Cardiol 50: 1852–1856. [DOI] [PubMed] [Google Scholar]
- Storey RF, James SK, Siegbahn A, Varenhorst C, Held C, Ycas J, et al. (2014). Lower mortality following pulmonary adverse events and sepsis with ticagrelor compared to clopidogrel in the PLATO study. Platelets 25: 517–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sürer S, Toktas F, Ay D, Eris C, Yavuz S, Turk T, et al. (2014). Effect of the P2Y12 antagonist Ticagrelor on neointimal hyperplasia in a rabbit carotid anastomosis model. Interact Cardiovasc Thorac Surg 19: 198–204. [DOI] [PubMed] [Google Scholar]
- Sugidachi A, Ogawa T, Kurihara A, Hagihara K, Jakubowski JA, Hashimoto M, et al. (2007). The greater in vivo antiplatelet effects of prasugrel as compared to clopidogrel reflect more efficient generation of its active metabolite with similar antiplatelet activity to that of clopidogrel's active metabolite. J Thromb Haemost 7: 1545–1551. [DOI] [PubMed] [Google Scholar]
- Takeda M, Yamashita T, Shinohara M, Sasaki N, Tawa H, Nakajima K, et al. (2012). Beneficial effect of anti‐platelet therapies on atherosclerotic lesion formation assessed by phase‐contrast X‐ray CT imaging. Int J Cardiovasc Imaging 28: 1181–1191. [DOI] [PubMed] [Google Scholar]
- Teng B, Smith JD, Rosenfeld ME, Robinet P, Davis ME, Morrison RR, et al. (2014). A₁ adenosine receptor deficiency or inhibition reduces atherosclerotic lesions in apolipoprotein E deficient mice. Cardiovasc Res 102: 157–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomas M, Judge H, Dockrell D, Sabroe I, Storey R (2014). Ticagrelor inhibits release of pro‐inflammatory cytokines TNF and IL‐6 during human endotoxaemia. Heart 100 (S3): A117–A118. [Google Scholar]
- Thomas MR, Storey RF (2015). Effect of P2Y12 inhibitors on inflammation and immunity. Thromb Haemost 114: 449–458. [DOI] [PubMed] [Google Scholar]
- Torngren K, Ohman J, Salmi H, Larsson J, Erlinge D (2013). Ticagrelor improves peripheral arterial function in patients with a previous acute coronary syndrome. Cardiology 124: 252–258. [DOI] [PubMed] [Google Scholar]
- Totani L, Dell'Elba G, Martelli N, Di Santo A, Piccoli A, Amore C, et al. (2012). Prasugrel inhibits platelet‐leukocyte interaction and reduces inflammatory markers in a model of endotoxic shock in the mouse. Thromb Haemost 107: 1130–1140. [DOI] [PubMed] [Google Scholar]
- van Giezen JJ, Nilsson L, Berntsson P, Wissing BM, Giordanetto F, Tomlinson W, et al. (2009). Ticagrelor binds to human P2Y(12) independently from ADP but antagonizes ADP‐induced receptor signaling and platelet aggregation. J Thromb Haemost 7: 1556–1565. [DOI] [PubMed] [Google Scholar]
- Varenhorst C, Alström U, Scirica BM, Hogue CW, Åsenblad N, Storey RF, et al. (2012). Factors contributing to the lower mortality with ticagrelor compared with clopidogrel in patients undergoing coronary artery bypass surgery. J Am Coll Cardiol 60: 1623–1630. [DOI] [PubMed] [Google Scholar]
- Vivekananthan DP, Bhatt DL, Chew DP, Zidar FJ, Chan AW, Moliterno DJ, et al. (2004). Effect of clopidogrel pretreatment on periprocedural rise in C‐reactive protein after percutaneous coronary intervention. Am J Cardiol 94: 358–360. [DOI] [PubMed] [Google Scholar]
- Waksman R, Pakala R, Roy P, Baffour R, Hellinga D, Seabron R, et al. (2008). Effect of clopidogrel on neointimal formation and inflammation in balloon‐denuded and radiated hypercholesterolemic rabbit iliac arteries. J Interv Cardiol 21: 122–128. [DOI] [PubMed] [Google Scholar]
- Wallentin L, Varenhorst C, James S, Erlinge D, Braun OO, Jakubowski JA, et al. (2008). Prasugrel achieves greater and faster P2Y12receptor‐mediated platelet inhibition than clopidogrel due to more efficient generation of its active metabolite in aspirin‐treated patients with coronary artery disease. Eur Heart J 29: 21–30. [DOI] [PubMed] [Google Scholar]
- Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, et al. (2009). Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 361: 1045–1057. [DOI] [PubMed] [Google Scholar]
- Wang L, Jacobsen SE, Bengtsson A, Erlinge D (2004). P2 receptor mRNA expression profiles in human lymphocytes, monocytes and CD34+ stem and progenitor cells. BMC Immunol 5: 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Zhang W, Zhu C, Bucher C, Blazar BR, Zhang C, et al. (2009). Inactivation of the adenosine A2A receptor protects apolipoprotein E‐deficient mice from atherosclerosis. Arterioscler Thromb Vasc Biol 29: 1046–1052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang K, Zhou X, Huang Y, Khalil M, Wiktor D, van Giezen JJ, et al. (2010). Adjunctive treatment with ticagrelor, but not clopidogrel, added to tPA enables sustained coronary artery recanalisation with recovery of myocardium perfusion in a canine coronary thrombosis model. Thromb Haemost 104: 609–617. [DOI] [PubMed] [Google Scholar]
- Warner TD, Nylander S, Whatling C (2011). Anti‐platelet therapy: cyclo‐oxygenase inhibition and the use of aspirin with particular regard to dual anti‐platelet therapy. Br. J Clin Pharmacol 72: 619–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Warnholtz A, Ostad MA, Velich N, Trautmann C, Schinzel R, Walter U, et al. (2008). A single loading dose of clopidogrel causes dose‐dependent improvement of endothelial dysfunction in patients with stable coronary artery disease: results of a double‐blind, randomized study. Atherosclerosis 196: 689–695. [DOI] [PubMed] [Google Scholar]
- Welsh RC, Rao SV, Zeymer U, Thompson VP, Huber K, Kochman J, et al. (2012). A randomized, double‐blind, active‐controlled phase 2 trial to evaluate a novel selective and reversible intravenous and oral P2Y12 inhibitor elinogrel versus clopidogrel in patients undergoing nonurgent percutaneous coronary intervention: the INNOVATE‐PCI trial. Circ Cardiovasc Interv 5: 336–346. [DOI] [PubMed] [Google Scholar]
- West LE, Steiner T, Judge HM, Francis SE, Storey RF (2014). Vessel wall, not platelet, P2Y12 potentiates early atherogenesis. Cardiovasc Res 102: 429–435. [DOI] [PubMed] [Google Scholar]
- Wihlborg AK, Wang L, Braun OO, Eyjolfsson A, Gustafsson R, Gudbjartsson T, et al. (2004). ADP receptor P2Y12 is expressed in vascular smooth muscle cells and stimulates contraction in human blood vessels. Arterioscler Thromb Vasc Biol 24: 1810–1815. [DOI] [PubMed] [Google Scholar]
- Willoughby SR, Luu LJ, Cameron JD, Nelson AJ, Schultz CD, Worthley SG, et al. (2014). Clopidogrel improves microvascular endothelial function in subjects with stable coronary artery disease. Heart Lung Circ 23: 534–541. [DOI] [PubMed] [Google Scholar]
- Wittfeldt A, Emanuelsson H, Brandrup‐Wognsen G, van Giezen JJ, Jonasson J, Nylander S, et al. (2013). Ticagrelor enhances adenosine‐induced coronary vasodilatory responses in humans. J Am Coll Cardiol 61: 723–727. [DOI] [PubMed] [Google Scholar]
- Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, et al. (2007). Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 357: 2001–2015. [DOI] [PubMed] [Google Scholar]
- Woo JS, Kim W, Jang HH, Kim JB, Kim WS, Kim KS (2014). Effect of platelet reactivity, endothelial function, and inflammatory status on outcomes in patients with stable angina pectoris on clopidogrel therapy. Am J Cardiol 113: 786–792. [DOI] [PubMed] [Google Scholar]
- Xiao Z, Théroux P (2004). Clopidogrel inhibits platelet‐leukocyte interactions and thrombin receptor agonist peptide‐induced platelet activation in patients with an acute coronary syndrome. J Am Coll Cardiol 43: 1982–1988. [DOI] [PubMed] [Google Scholar]
- Yang XM, Liu Y, Cui L, Yang X, Liu Y, Tandon N, et al. (2013). Platelet P2Y₁₂ blockers confer direct postconditioning‐like protection in reperfused rabbit hearts. J Cardiovasc Pharmacol Ther 18: 251–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yeh ET, Khan BV (2006). The potential role of antiplatelet agents in modulating inflammatory markers in atherothrombosis. J Thromb Haemost 4: 2308–2316. [DOI] [PubMed] [Google Scholar]
- Ye Y, Birnbaum GD, Perez‐Polo JR, Nanhwan MK, Nylander S, Birnbaum Y (2015). Ticagrelor Protects the Heart Against Reperfusion Injury and Improves Remodeling After Myocardial Infarction. Arterioscler Thromb Vasc Biol 35: 1805–1814. [DOI] [PubMed] [Google Scholar]
- Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK (2001). Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial Investigators. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST‐segment elevation. N Engl J Med 345: 494–502. [DOI] [PubMed] [Google Scholar]